Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.467
Filter
1.
Nat Commun ; 15(1): 6740, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39112531

ABSTRACT

Glioblastoma (GBM) is the most common brain tumor and remains incurable. Primary GBM cultures are widely used tools for drug screening, but there is a lack of genomic and pharmacological characterization for these primary GBM cultures. Here, we collect 50 patient-derived glioma cell (PDGC) lines and characterize them by whole genome sequencing, RNA sequencing, and drug response screening. We identify three molecular subtypes among PDGCs: mesenchymal (MES), proneural (PN), and oxidative phosphorylation (OXPHOS). Drug response profiling reveals that PN subtype PDGCs are sensitive to tyrosine kinase inhibitors, whereas OXPHOS subtype PDGCs are sensitive to histone deacetylase inhibitors, oxidative phosphorylation inhibitors, and HMG-CoA reductase inhibitors. PN and OXPHOS subtype PDGCs stably form tumors in vivo upon intracranial transplantation into immunodeficient mice, whereas most MES subtype PDGCs fail to form tumors in vivo. In addition, PDGCs cultured by serum-free medium, especially long-passage PDGCs, carry MYC/MYCN amplification, which is rare in GBM patients. Our study provides a valuable resource for understanding primary glioma cell cultures and clinical translation and highlights the problems of serum-free PDGC culture systems that cannot be ignored.


Subject(s)
Brain Neoplasms , Glioma , Humans , Animals , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Mice , Glioma/genetics , Glioma/pathology , Glioma/drug therapy , Glioma/metabolism , Oxidative Phosphorylation/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/drug therapy , Glioblastoma/metabolism , Female , Male , Whole Genome Sequencing , Xenograft Model Antitumor Assays , Genomics/methods , Gene Expression Regulation, Neoplastic/drug effects , Multiomics
2.
Cell Death Dis ; 15(8): 582, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122684

ABSTRACT

Age-related macular degeneration (AMD) causes severe blindness in the elderly due to choroidal neovascularization (CNV), which results from the dysfunction of the retinal pigment epithelium (RPE). While normal RPE depends exclusively on mitochondrial oxidative phosphorylation for energy production, the inflammatory conditions associated with metabolic reprogramming of the RPE play a pivotal role in CNV. Although mitochondrial pyruvate dehydrogenase kinase (PDK) is a central node of energy metabolism, its role in the development of CNV in neovascular AMD has not been investigated. In the present study, we used a laser-induced CNV mouse model to evaluate the effects of Pdk4 gene ablation and treatment with pan-PDK or specific PDK4 inhibitors on fluorescein angiography and CNV lesion area. Among PDK isoforms, only PDK4 was upregulated in the RPE of laser-induced CNV mice, and Pdk4 gene ablation attenuated CNV. Next, we evaluated mitochondrial changes mediated by PDK1-4 inhibition using siRNA or PDK inhibitors in inflammatory cytokine mixture (ICM)-treated primary human RPE (hRPE) cells. PDK4 silencing only in ICM-treated hRPE cells restored mitochondrial respiration and reduced inflammatory cytokine secretion. Likewise, GM10395, a specific PDK4 inhibitor, restored oxidative phosphorylation and decreased ICM-induced upregulation of inflammatory cytokine secretion. In a laser-induced CNV mouse model, GM10395 significantly alleviated CNV. Taken together, we demonstrate that specific PDK4 inhibition could be a therapeutic strategy for neovascular AMD by preventing mitochondrial metabolic reprogramming in the RPE under inflammatory conditions.


Subject(s)
Choroidal Neovascularization , Disease Models, Animal , Macular Degeneration , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Retinal Pigment Epithelium , Animals , Humans , Mice , Macular Degeneration/metabolism , Macular Degeneration/pathology , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Choroidal Neovascularization/drug therapy , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/drug effects , Oxidative Phosphorylation/drug effects , Metabolic Reprogramming
3.
Int J Mol Sci ; 25(15)2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39126062

ABSTRACT

Little is known about the effects of statins, which are cholesterol-lowering drugs, on the bioenergetic functions of mitochondria in the brain. This study aimed to elucidate the direct effects of atorvastatin and simvastatin on the bioenergetics of isolated rat brain mitochondria by measuring the statin-induced changes in respiratory chain activity, ATP synthesis efficiency, and the production of reactive oxygen species (ROS). Our results in isolated brain mitochondria are the first to demonstrate that atorvastatin and simvastatin dose-dependently significantly inhibit the activity of the mitochondrial respiratory chain, resulting in a decreased respiratory rate, a decreased membrane potential, and increased ROS formation. Moreover, the tested statins reduced mitochondrial coupling parameters, the ADP/O ratio, the respiratory control ratio, and thus, the oxidative phosphorylation efficiency in brain mitochondria. Among the oxidative phosphorylation complexes, statin-induced mitochondrial impairment concerned complex I, complex III, and ATP synthase activity. The calcium-containing atorvastatin had a significantly more substantial effect on isolated brain mitochondria than simvastatin. The higher inhibitory effect of atorvastatin was dependent on calcium ions, which may lead to the disruption of calcium homeostasis in mitochondria. These findings suggest that while statins are effective in their primary role as cholesterol-lowering agents, their use may impair mitochondrial function, which may have consequences for brain health, particularly when mitochondrial energy efficiency is critical.


Subject(s)
Atorvastatin , Brain , Energy Metabolism , Mitochondria , Reactive Oxygen Species , Simvastatin , Animals , Atorvastatin/pharmacology , Simvastatin/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Brain/drug effects , Brain/metabolism , Rats , Energy Metabolism/drug effects , Reactive Oxygen Species/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Oxidative Phosphorylation/drug effects , Adenosine Triphosphate/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Calcium/metabolism
4.
J Transl Med ; 22(1): 757, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39135106

ABSTRACT

BACKGROUND: Multi-drug resistance of poly(morpho)nuclear giant cells (PGCs) determines their cytoprotective and generative potential in cancer ecosystems. However, mechanisms underlying the involvement of PGCs in glioblastoma multiforme (GBM) adaptation to chemotherapeutic regimes remain largely obscure. In particular, metabolic reprogramming of PGCs has not yet been considered in terms of GBM recovery from doxorubicin (DOX)-induced stress. METHODS: Long-term proteomic and metabolic cell profiling was applied to trace the phenotypic dynamics of GBM populations subjected to pulse DOX treatment in vitro, with a particular focus on PGC formation and its metabolic background. The links between metabolic reprogramming, drug resistance and drug retention capacity of PGCs were assessed, along with their significance for GBM recovery from DOX-induced stress. RESULTS: Pulse DOX treatment triggered the transient formation of PGCs, followed by the appearance of small expanding cell (SEC) clusters. Development of PGCs was accompanied by the mobilization of their metabolic proteome, transient induction of oxidative phosphorylation (OXPHOS), and differential intracellular accumulation of NADH, NADPH, and ATP. The metabolic background of PGC formation was confirmed by the attenuation of GBM recovery from DOX-induced stress following the chemical inhibition of GSK-3ß, OXPHOS, and the pentose phosphate pathway. Concurrently, the mobilization of reactive oxygen species (ROS) scavenging systems and fine-tuning of NADPH-dependent ROS production systems in PGCs was observed. These processes were accompanied by perinuclear mobilization of ABCB1 and ABCG2 transporters and DOX retention in the perinuclear PGC compartments. CONCLUSIONS: These data demonstrate the cooperative pattern of GBM recovery from DOX-induced stress and the crucial role of metabolic reprogramming of PGCs in this process. Metabolic reprogramming enhances the efficiency of self-defense systems and increases the DOX retention capacity of PGCs, potentially reducing DOX bioavailability in the proximity of SECs. Consequently, the modulation of PGC metabolism is highlighted as a potential target for intervention in glioblastoma treatment.


Subject(s)
Doxorubicin , Glioblastoma , Glioblastoma/pathology , Glioblastoma/metabolism , Humans , Doxorubicin/pharmacology , Cell Line, Tumor , Stress, Physiological/drug effects , Cellular Reprogramming/drug effects , Cell Nucleus/metabolism , Cell Nucleus/drug effects , Proteomics , Drug Resistance, Neoplasm/drug effects , Oxidative Phosphorylation/drug effects , Metabolic Reprogramming
5.
Toxins (Basel) ; 16(7)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39057927

ABSTRACT

In this paper, we provide an overview of mitochondrial bioenergetics and specific conditions that lead to the formation of non-bilayer structures in mitochondria. Secondly, we provide a brief overview on the structure/function of cytotoxins and how snake venom cytotoxins have contributed to increasing our understanding of ATP synthesis via oxidative phosphorylation in mitochondria, to reconcile some controversial aspects of the chemiosmotic theory. Specifically, we provide an emphasis on the biochemical contribution of delocalized and localized proton movement, involving direct transport of protons though the Fo unit of ATP synthase or via the hydrophobic environment at the center of the inner mitochondrial membrane (proton circuit) on oxidative phosphorylation, and how this influences the rate of ATP synthesis. Importantly, we provide new insights on the molecular mechanisms through which cobra venom cytotoxins affect mitochondrial ATP synthesis, mitochondrial structure, and dynamics. Finally, we provide a perspective for the use of cytotoxins as novel pharmacological tools to study membrane bioenergetics and mitochondrial biology, how they can be used in translational research, and their potential therapeutic applications.


Subject(s)
Elapid Venoms , Energy Metabolism , Mitochondria , Mitochondrial Membranes , Animals , Energy Metabolism/drug effects , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Humans , Elapid Venoms/chemistry , Elapid Venoms/toxicity , Elapid Venoms/metabolism , Cytotoxins/pharmacology , Cytotoxins/toxicity , Cytotoxins/chemistry , Adenosine Triphosphate/metabolism , Oxidative Phosphorylation/drug effects
6.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-39000550

ABSTRACT

The effect of the modulators of the mitochondrial ATP-dependent potassium channel (mitoKATP) on the structural and biochemical alterations in the substantia nigra and brain tissues was studied in a rat model of Parkinson's disease induced by rotenone. It was found that, in experimental parkinsonism accompanied by characteristic motor deficits, both neurons and the myelin sheath of nerve fibers in the substantia nigra were affected. Changes in energy and ion exchange in brain mitochondria were also revealed. The nucleoside uridine, which is a source for the synthesis of the mitoKATP channel opener uridine diphosphate, was able to dose-dependently decrease behavioral disorders and prevent the death of animals, which occurred for about 50% of animals in the model. Uridine prevented disturbances in redox, energy, and ion exchanges in brain mitochondria, and eliminated alterations in their structure and the myelin sheath in the substantia nigra. Cytochemical examination showed that uridine restored the indicators of oxidative phosphorylation and glycolysis in peripheral blood lymphocytes. The specific blocker of the mitoKATP channel, 5-hydroxydecanoate, eliminated the positive effects of uridine, suggesting that this channel is involved in neuroprotection. Taken together, these findings indicate the promise of using the natural metabolite uridine as a new drug to prevent and, possibly, stop the progression of Parkinson's disease.


Subject(s)
Mitochondria , Potassium Channels , Rotenone , Uridine , Animals , Uridine/pharmacology , Uridine/metabolism , Rats , Potassium Channels/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Male , Disease Models, Animal , Parkinson Disease/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/etiology , Parkinson Disease/pathology , Substantia Nigra/metabolism , Substantia Nigra/drug effects , Substantia Nigra/pathology , Neuroprotective Agents/pharmacology , Oxidative Phosphorylation/drug effects , Rats, Wistar , Decanoic Acids/pharmacology , Hydroxy Acids/pharmacology
7.
Chem Biol Interact ; 399: 111141, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-38992767

ABSTRACT

Mitochondrial Pyruvate Carrier 1 (MPC1) is localized on mitochondrial outer membrane to mediate the transport of pyruvate from cytosol to mitochondria. It is also well known to act as a tumor suppressor. Hexavalent chromium (Cr (VI)) contamination poses a global challenge due to its high toxicity and carcinogenesis. This research was intended to probe the potential mechanism of MPC1 in the effect of Cr (VI)-induced carcinogenesis. First, Cr (VI)-treatments decreased the expression of MPC1 in vitro and in vivo. Overexpression of MPC1 inhibited Cr (VI)-induced glycolysis and migration in A549 cells. Then, high mobility group A2 (HMGA2) protein strongly suppressed the transcription of MPC1 by binding to its promoter, and HMGA2/MPC1 axis played an important role in oxidative phosphorylation (OXPHOS), glycolysis and cell migration. Furthermore, endoplasmic reticulum (ER) stress made a great effect on the interaction between HMGA2 and MPC1. Finally, the mammalian target of the rapamycin (mTOR) was determined to mediate MPC1-regulated OXPHOS, aerobic glycolysis and cell migration. Collectively, our data revealed a novel HMGA2/MPC-1/mTOR signaling pathway to promote cell growth via facilitating the metabolism reprogramming from OXPHOS to aerobic glycolysis, which might be a potential therapy for cancers.


Subject(s)
Cell Movement , Cell Proliferation , Chromium , Glycolysis , HMGA2 Protein , Monocarboxylic Acid Transporters , Signal Transduction , TOR Serine-Threonine Kinases , Humans , TOR Serine-Threonine Kinases/metabolism , Glycolysis/drug effects , Signal Transduction/drug effects , HMGA2 Protein/metabolism , HMGA2 Protein/genetics , Cell Movement/drug effects , Chromium/pharmacology , Cell Proliferation/drug effects , Animals , Monocarboxylic Acid Transporters/metabolism , Monocarboxylic Acid Transporters/antagonists & inhibitors , A549 Cells , Mice , Endoplasmic Reticulum Stress/drug effects , Mice, Nude , Membrane Transport Proteins/metabolism , Oxidative Phosphorylation/drug effects , Mice, Inbred BALB C , Cell Line, Tumor , Mitochondrial Membrane Transport Proteins
8.
Proc Natl Acad Sci U S A ; 121(30): e2321972121, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39008677

ABSTRACT

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection inhibits mitochondrial oxidative phosphorylation (OXPHOS) and elevates mitochondrial reactive oxygen species (ROS, mROS) which activates hypoxia-inducible factor-1alpha (HIF-1α), shifting metabolism toward glycolysis to drive viral biogenesis but also causing the release of mitochondrial DNA (mtDNA) and activation of innate immunity. To determine whether mitochondrially targeted antioxidants could mitigate these viral effects, we challenged mice expressing human angiotensin-converting enzyme 2 (ACE2) with SARS-CoV-2 and intervened using transgenic and pharmacological mitochondrially targeted catalytic antioxidants. Transgenic expression of mitochondrially targeted catalase (mCAT) or systemic treatment with EUK8 decreased weight loss, clinical severity, and circulating levels of mtDNA; as well as reduced lung levels of HIF-1α, viral proteins, and inflammatory cytokines. RNA-sequencing of infected lungs revealed that mCAT and Eukarion 8 (EUK8) up-regulated OXPHOS gene expression and down-regulated HIF-1α and its target genes as well as innate immune gene expression. These data demonstrate that SARS-CoV-2 pathology can be mitigated by catalytically reducing mROS, potentially providing a unique host-directed pharmacological therapy for COVID-19 which is not subject to viral mutational resistance.


Subject(s)
Antioxidants , COVID-19 , Mice, Transgenic , Mitochondria , Oxidative Phosphorylation , SARS-CoV-2 , Animals , Mice , COVID-19/virology , COVID-19/metabolism , COVID-19/immunology , COVID-19/pathology , Antioxidants/metabolism , Antioxidants/pharmacology , Mitochondria/metabolism , Mitochondria/drug effects , SARS-CoV-2/drug effects , Oxidative Phosphorylation/drug effects , Humans , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Lung/virology , Lung/pathology , Lung/metabolism , Reactive Oxygen Species/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Catalase/metabolism , Catalase/genetics , COVID-19 Drug Treatment , Disease Models, Animal , Immunity, Innate
9.
Nat Commun ; 15(1): 5759, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982116

ABSTRACT

Type I interferons have been well recognized for their roles in various types of immune cells during tumor immunotherapy. However, their direct effects on tumor cells are less understood. Oxidative phosphorylation is typically latent in tumor cells. Whether oxidative phosphorylation can be targeted for immunotherapy remains unclear. Here, we find that tumor cell responsiveness to type I, but not type II interferons, is essential for CD47-SIRPα blockade immunotherapy in female mice. Mechanistically, type I interferons directly reprogram tumor cell metabolism by activating oxidative phosphorylation for ATP production in an ISG15-dependent manner. ATP extracellular release is also promoted by type I interferons due to enhanced secretory autophagy. Functionally, tumor cells with genetic deficiency in oxidative phosphorylation or autophagy are resistant to CD47-SIRPα blockade. ATP released upon CD47-SIRPα blockade is required for antitumor T cell response induction via P2X7 receptor-mediated dendritic cell activation. Based on this mechanism, combinations with inhibitors of ATP-degrading ectoenzymes, CD39 and CD73, are designed and show synergistic antitumor effects with CD47-SIRPα blockade. Together, these data reveal an important role of type I interferons on tumor cell metabolic reprograming for tumor immunotherapy and provide rational strategies harnessing this mechanism for enhanced efficacy of CD47-SIRPα blockade.


Subject(s)
Adenosine Triphosphate , CD47 Antigen , Interferon Type I , Oxidative Phosphorylation , Receptors, Immunologic , Signal Transduction , Animals , CD47 Antigen/metabolism , CD47 Antigen/genetics , Interferon Type I/metabolism , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Female , Mice , Adenosine Triphosphate/metabolism , Oxidative Phosphorylation/drug effects , Cell Line, Tumor , Mice, Inbred C57BL , Immunotherapy/methods , Humans , Dendritic Cells/immunology , Dendritic Cells/metabolism , Receptors, Purinergic P2X7/metabolism , Receptors, Purinergic P2X7/genetics , Autophagy/drug effects , Apyrase/metabolism , Mice, Knockout , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Cytokines/metabolism
10.
J Pineal Res ; 76(5): e12991, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39039850

ABSTRACT

Although rapid progression and a poor prognosis in influenza A virus (IAV) infection-induced acute exacerbation of chronic obstructive pulmonary disease (AECOPD) are frequently associated with metabolic energy disorders, the underlying mechanisms and rescue strategies remain unknown. We herein demonstrated that the level of resting energy expenditure increased significantly in IAV-induced AECOPD patients and that cellular energy exhaustion emerged earlier and more significantly in IAV-infected primary COPD bronchial epithelial (pDHBE) cells. The differentially expressed genes were enriched in the oxidative phosphorylation (OXPHOS) pathway; additionally, we consistently uncovered much earlier ATP exhaustion, more severe mitochondrial structural destruction and dysfunction, and OXPHOS impairment in IAV-inoculated pDHBE cells, and these changes were rescued by melatonin. The level of OMA1-dependent cleavage of OPA1 in the mitochondrial inner membrane and the shift in energy metabolism from OXPHOS to glycolysis were significantly increased in IAV-infected pDHBE cells; however, these changes were rescued by OMA1-siRNA or melatonin further treatment. Collectively, our data revealed that melatonin rescued IAV-induced cellular energy exhaustion via OMA1-OPA1-S to improve the clinical prognosis in COPD. This treatment may serve as a potential therapeutic agent for patients in which AECOPD is induced by IAV.


Subject(s)
Energy Metabolism , GTP Phosphohydrolases , Influenza A virus , Melatonin , Pulmonary Disease, Chronic Obstructive , Humans , Energy Metabolism/drug effects , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/genetics , Influenza A virus/drug effects , Influenza, Human/metabolism , Influenza, Human/drug therapy , Melatonin/pharmacology , Metalloendopeptidases , Oxidative Phosphorylation/drug effects , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/drug therapy
11.
Cell Death Dis ; 15(7): 475, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961053

ABSTRACT

Deregulated apoptosis signaling is characteristic for many cancers and contributes to leukemogenesis and treatment failure in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Apoptosis is controlled by different pro- and anti-apoptotic molecules. Inhibition of anti-apoptotic molecules like B-cell lymphoma 2 (BCL-2) has been developed as therapeutic strategy. Venetoclax (VEN), a selective BCL-2 inhibitor has shown clinical activity in different lymphoid malignancies and is currently evaluated in first clinical trials in BCP-ALL. However, insensitivity to VEN has been described constituting a major clinical concern. Here, we addressed and modeled VEN-resistance in BCP-ALL, investigated the underlying mechanisms in cell lines and patient-derived xenograft (PDX) samples and identified potential strategies to overcome VEN-insensitivity. Leukemia lines with VEN-specific resistance were generated in vitro and further characterized using RNA-seq analysis. Interestingly, gene sets annotated to the citric/tricarboxylic acid cycle and the respiratory electron transport chain were significantly enriched and upregulated, indicating increased mitochondrial metabolism in VEN-resistant ALL. Metabolic profiling showed sustained high mitochondrial metabolism in VEN-resistant lines as compared to control lines. Accordingly, primary PDX-ALL samples with intrinsic VEN-insensitivity showed higher oxygen consumption and ATP production rates, further highlighting that increased mitochondrial activity is a characteristic feature of VEN-resistant ALL. VEN-resistant PDX-ALL showed significant higher mitochondrial DNA content and differed in mitochondria morphology with significantly larger and elongated structures, further corroborating our finding of augmented mitochondrial metabolism upon VEN-resistance. Using Oligomycin, an inhibitor of the complex V/ATPase subunit, we found synergistic activity and apoptosis induction in VEN-resistant BCP-ALL cell lines and PDX samples, demonstrating that acquired and intrinsic VEN-insensitivity can be overcome by co-targeting BCL-2 and the OxPhos pathway. These findings of reprogrammed, high mitochondrial metabolism in VEN-resistance and synergistic activity upon co-targeting BCL-2 and oxidative phosphorylation strongly suggest further preclinical and potential clinical evaluation in VEN-resistant BCP-ALL.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic , Drug Resistance, Neoplasm , Mitochondria , Oxidative Phosphorylation , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Sulfonamides , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Humans , Oxidative Phosphorylation/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Sulfonamides/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Cell Line, Tumor , Mice , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Xenograft Model Antitumor Assays , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics
12.
Acta Biomater ; 184: 368-382, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908417

ABSTRACT

Starvation therapy aims to "starve" tumor cells by cutting off their nutritional supply. However, due to the complex and varied energy metabolism of tumors, targeting a single nutrient supply often fails to yield significant therapeutic benefits. This study proposes a tumor energy cocktail therapy that combines metformin, an oxidative phosphorylation inhibitor, with 2-deoxy-d-glucose (2-DG), a glycolysis inhibitor, to target tumor cells. To minimize the dosage of both drugs, we have developed a drug delivery strategy that prepared metformin as a nanoderivative, denoted as MA-dots. These MA-dots not only preserve the antitumor properties of metformin but also serve as a targeted delivery platform for 2-DG, ensuring its direct reach to the tumor site. Upon reaching the acidic tumor environment, the composite disintegrates, releasing 2-DG to inhibit glycolysis by targeting hexokinase 2 (HK2), the key enzyme in glycolysis, while MA-dots inhibit mitochondrial OXPHOS. This dual action significantly reduces ATP production in tumor cells, leading to apoptosis. In human lung tumor cells, the half-maximal inhibitory concentration (IC50) of 2-DG@MA-dots was significantly lower than that of either metformin or 2-DG alone, showing a nearly 100-fold and 30-fold reduction in IC50 values to 11.78 µg mL-1, from 1159 µg mL-1 and 351.20 µg mL-1, respectively. In studies with A549 tumor-bearing mice, the combination of low-dose 2-DG and metformin did not impede tumor growth, whereas 2-DG@MA-dots markedly decreased tumor volume, with the mean final tumor volume in the combination treatment group being approximately 89 times greater than that in the 2-DG@MA-dot group. STATEMENT OF SIGNIFICANCE: Metformin is a promising antitumor agent capable of modulating mitochondrial oxidative phosphorylation to inhibit cancer growth. However, its antitumor efficacy is limited when used alone due to compensatory energy mechanisms. Hence, we introduced glycolysis inhibitor 2-deoxy-d-glucose (2-DG) to inhibit an alternative tumor energy pathway. In our study, we developed a drug delivery strategy using metformin-derived nanomedicine (MA-dots) to load 2-DG. This approach enables the co-delivery of both drugs and their synergistic effect at the tumor site, disrupting both energy pathways and introducing an innovative "energy cocktail therapy".


Subject(s)
Deoxyglucose , Metformin , Humans , Deoxyglucose/pharmacology , Metformin/pharmacology , Metformin/therapeutic use , Animals , Mice , Mice, Nude , Energy Metabolism/drug effects , Glycolysis/drug effects , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Hexokinase/metabolism , Cell Line, Tumor , Apoptosis/drug effects , Oxidative Phosphorylation/drug effects , A549 Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry
13.
Neurobiol Dis ; 199: 106573, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38901783

ABSTRACT

Arketamine, the (R)-enantiomer of ketamine, exhibits antidepressant-like effects in mice, though the precise molecular mechanisms remain elusive. It has been shown to reduce splenomegaly and depression-like behaviors in the chronic social defeat stress (CSDS) model of depression. This study investigated whether the spleen contributes to the antidepressant-like effects of arketamine in the CSDS model. We found that splenectomy significantly inhibited arketamine's antidepressant-like effects in CSDS-susceptible mice. RNA-sequencing analysis identified the oxidative phosphorylation (OXPHOS) pathway in the prefrontal cortex (PFC) as a key mediator of splenectomy's impact on arketamine's effects. Furthermore, oligomycin A, an inhibitor of the OXPHOS pathway, reversed the suppressive effects of splenectomy on arketamine's antidepressant-like effects. Specific genes within the OXPHOS pathways, such as COX11, UQCR11 and ATP5e, may contribute to these inhibitory effects. Notably, transforming growth factor (TGF)-ß1, along with COX11, appears to modulate the suppressive effects of splenectomy and contribute to arketamine's antidepressant-like effects. Additionally, SRI-01138, an agonist of the TGF-ß1 receptor, alleviated the inhibitory effects of splenectomy on arketamine's antidepressant-like effects. Subdiaphragmatic vagotomy also counteracted the inhibitory effects of splenectomy on arketamine's antidepressant-like effects in CSDS-susceptible mice. These findings suggest that the OXPHOS pathway and TGF-ß1 in the PFC play significant roles in the antidepressant-like effects of arketamine, mediated through the spleen-brain axis via the vagus nerve.


Subject(s)
Antidepressive Agents , Ketamine , Mice, Inbred C57BL , Oxidative Phosphorylation , Spleen , Splenectomy , Vagus Nerve , Animals , Ketamine/pharmacology , Antidepressive Agents/pharmacology , Spleen/drug effects , Spleen/metabolism , Mice , Male , Vagus Nerve/drug effects , Vagus Nerve/metabolism , Oxidative Phosphorylation/drug effects , Depression/drug therapy , Depression/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Brain/drug effects , Brain/metabolism , Stress, Psychological/metabolism , Stress, Psychological/drug therapy , Social Defeat
14.
Nano Lett ; 24(27): 8257-8267, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38920296

ABSTRACT

Osseointegration is the most important factor determining implant success. The surface modification of TiO2 nanotubes prepared by anodic oxidation has remarkable advantages in promoting bone formation. However, the mechanism behind this phenomenon is still unintelligible. Here we show that the nanomorphology exhibited open and clean nanotube structure and strong hydrophilicity, and the nanomorphology significantly facilitated the adhesion, proliferation, and osteogenesis differentiation of stem cells. Exploring the mechanism, we found that the nanomorphology can enhance mitochondrial oxidative phosphorylation (OxPhos) by activating Piezo1 and increasing intracellular Ca2+. The increase in OxPhos can significantly uplift the level of acetyl-CoA in the cytoplasm but not significantly raise the level of acetyl-CoA in the nucleus, which was beneficial for the acetylation and stability of ß-catenin and ultimately promoted osteogenesis. This study provides a new interpretation for the regulatory mechanism of stem cell osteogenesis by nanomorphology.


Subject(s)
Cell Differentiation , Ion Channels , Osteogenesis , Surface Properties , Titanium , beta Catenin , Osteogenesis/drug effects , Titanium/chemistry , Titanium/pharmacology , beta Catenin/metabolism , Ion Channels/metabolism , Cell Differentiation/drug effects , Animals , Cell Proliferation/drug effects , Osseointegration/drug effects , Mice , Nanopores , Nanotubes/chemistry , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Oxidative Phosphorylation/drug effects , Prostheses and Implants , Cell Adhesion/drug effects
15.
Sci Transl Med ; 16(751): eadi5336, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38865484

ABSTRACT

In chronic myeloid leukemia (CML), the persistence of leukemic stem cells (LSCs) after treatment with tyrosine kinase inhibitors (TKIs), such as imatinib, can lead to disease relapse. It is known that therapy-resistant LSCs rely on oxidative phosphorylation (OXPHOS) for their survival and that targeting mitochondrial respiration sensitizes CML LSCs to imatinib treatment. However, current OXPHOS inhibitors have demonstrated limited efficacy or have shown adverse effects in clinical trials, highlighting that identification of clinically safe oxidative pathway inhibitors is warranted. We performed a high-throughput drug repurposing screen designed to identify mitochondrial metabolism inhibitors in myeloid leukemia cells. This identified lomerizine, a US Food and Drug Administration (FDA)-approved voltage-gated Ca2+ channel blocker now used for the treatment of migraines, as one of the top hits. Transcriptome analysis revealed increased expression of voltage-gated CACNA1D and receptor-activated TRPC6 Ca2+ channels in CML LSCs (CD34+CD38-) compared with normal counterparts. This correlated with increased endoplasmic reticulum (ER) mass and increased ER and mitochondrial Ca2+ content in CML stem/progenitor cells. We demonstrate that lomerizine-mediated inhibition of Ca2+ uptake leads to ER and mitochondrial Ca2+ depletion, with similar effects seen after CACNA1D and TRPC6 knockdown. Through stable isotope-assisted metabolomics and functional assays, we observe that lomerizine treatment inhibits mitochondrial isocitrate dehydrogenase activity and mitochondrial oxidative metabolism and selectively sensitizes CML LSCs to imatinib treatment. In addition, combination treatment with imatinib and lomerizine reduced CML tumor burden, targeted CML LSCs, and extended survival in xenotransplantation model of human CML, suggesting this as a potential therapeutic strategy to prevent disease relapse in patients.


Subject(s)
Drug Repositioning , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Mitochondria , Humans , Mitochondria/metabolism , Mitochondria/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Animals , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/drug effects , Piperazines/pharmacology , Piperazines/therapeutic use , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Calcium/metabolism , Oxidative Phosphorylation/drug effects , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use
16.
mBio ; 15(7): e0129224, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38860823

ABSTRACT

Sepsis and chronic infections with Pseudomonas aeruginosa, a leading "ESKAPE" bacterial pathogen, are associated with increased morbidity and mortality and skeletal muscle atrophy. The actions of this pathogen on skeletal muscle remain poorly understood. In skeletal muscle, mitochondria serve as a crucial energy source, which may be perturbed by infection. Here, using the well-established backburn and infection model of murine P. aeruginosa infection, we deciphered the systemic impact of the quorum-sensing transcription factor MvfR (multiple virulence factor regulator) by interrogating, 5 days post-infection, its effect on mitochondrial-related functions in the gastrocnemius skeletal muscle and the outcome of the pharmacological inhibition of MvfR function and that of the mitochondrial-targeted peptide, Szeto-Schiller 31 (SS-31). Our findings show that the MvfR perturbs adenosine triphosphate generation, oxidative phosphorylation, and antioxidant response, elevates the production of reactive oxygen species, and promotes oxidative damage of mitochondrial DNA in the gastrocnemius muscle of infected mice. These impairments in mitochondrial-related functions were corroborated by the alteration of key mitochondrial proteins involved in electron transport, mitochondrial biogenesis, dynamics and quality control, and mitochondrial uncoupling. Pharmacological inhibition of MvfR using the potent anti-MvfR lead, D88, we developed, or the mitochondrial-targeted peptide SS-31 rescued the MvfR-mediated alterations observed in mice infected with the wild-type strain PA14. Our study provides insights into the actions of MvfR in orchestrating mitochondrial dysfunction in the skeletal murine muscle, and it presents novel therapeutic approaches for optimizing clinical outcomes in affected patients. IMPORTANCE: Skeletal muscle, pivotal for many functions in the human body, including breathing and protecting internal organs, contains abundant mitochondria essential for maintaining cellular homeostasis during infection. The effect of Pseudomonas aeruginosa (PA) infections on skeletal muscle remains poorly understood. Our study delves into the role of a central quorum-sensing transcription factor, multiple virulence factor regulator (MvfR), that controls the expression of multiple acute and chronic virulence functions that contribute to the pathogenicity of PA. The significance of our study lies in the role of MvfR in the metabolic perturbances linked to mitochondrial functions in skeletal muscle and the effectiveness of the novel MvfR inhibitor and the mitochondrial-targeted peptide SS-31 in alleviating the mitochondrial disturbances caused by PA in skeletal muscle. Inhibiting MvfR or interfering with its effects can be a potential therapeutic strategy to curb PA virulence.


Subject(s)
Bacterial Proteins , Muscle, Skeletal , Pseudomonas Infections , Pseudomonas aeruginosa , Quorum Sensing , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/physiology , Animals , Mice , Muscle, Skeletal/microbiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Pseudomonas Infections/microbiology , Pseudomonas Infections/drug therapy , Quorum Sensing/drug effects , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Disease Models, Animal , Virulence Factors/metabolism , Virulence Factors/genetics , Male , Oxidative Phosphorylation/drug effects , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/drug effects , Mice, Inbred C57BL , Oligopeptides/pharmacology , Adenosine Triphosphate/metabolism , Anti-Bacterial Agents/pharmacology
17.
Cell Physiol Biochem ; 58(3): 226-249, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38857359

ABSTRACT

BACKGROUND/AIMS: Important benefits of intermittent hypoxic training (IHT) have emerged as an effective tool for enhancing adaptive potential in different pathological states, among which acute hypoxia dominates. Therefore, the aim of our study was to evaluate the mechanisms related to the effects of the nitric oxide system (nitrites, nitrates, carbamide, and total polyamine content) on ADP-stimulated oxygen consumption and oxidative phosphorylation in heart and liver mitochondria and biomarkers of oxidative stress in the blood, heart, and liver of rats exposed to the IHT method and acute hypoxia and treated with the amino acid L-arginine (600 mg/kg, 30 min) or the NO synthase inhibitor L-NNA (35 mg/kg, 30 min) prior to each IHT session. METHODS: We analysed the modulation of the system of oxygen-dependent processes (mitochondrial respiration with the oxygraphic method, microsomal oxidation, and lipoperoxidation processes using biochemical methods) in tissues during IHT in the formation of short-term and long-term effects (30, 60, and 180 days after the last IHT session) with simultaneous administration of L-arginine. In particular, we investigated how mitochondrial functions are modulated during intermittent hypoxia with the use of oxidation substrates (succinate or α-ketoglutarate) in bioenergetic mechanisms of cellular stability and adaptation. RESULTS: The IHT method is associated with a significant increase in the production of endogenous nitric oxide measured by the levels of its stable metabolite, nitrite anion, in both plasma (almost 7-fold) and erythrocytes (more than 7-fold) of rats. The intensification of nitric oxide-dependent pathways of metabolic transformations in the energy supply processes in the heart and liver, accompanied by oscillatory mechanisms of adaptation in the interval mode, causes a probable decrease in the production of urea and polyamines in plasma and liver, but not in erythrocytes. The administration of L-arginine prior to the IHT sessions increased the level of the nitrite-reducing component of the nitric oxide cycle, which persisted for up to 180 days of the experiment. CONCLUSION: Thus, the efficacy of IHT and its nitrite-dependent component shown in this study is associated with the formation of long-term adaptive responses by preventing the intensification of lipoperoxidation processes in tissues due to pronounced changes in the main enzymes of antioxidant defence and stabilisation of erythrocyte membranes, which has a pronounced protective effect on the system of regulation of oxygen-dependent processes as a whole.


Subject(s)
Arginine , Hypoxia , Oxygen Consumption , Rats, Wistar , Animals , Male , Hypoxia/metabolism , Rats , Arginine/pharmacology , Arginine/analogs & derivatives , Arginine/metabolism , Oxygen Consumption/drug effects , Oxidative Stress/drug effects , Nitric Oxide/metabolism , Oxygen/metabolism , Adaptation, Physiological , Mitochondria, Liver/metabolism , Mitochondria, Liver/drug effects , Oxidative Phosphorylation/drug effects , Liver/metabolism , Liver/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Lipid Peroxidation/drug effects , Nitrites/metabolism
18.
Molecules ; 29(12)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38930984

ABSTRACT

Halogenated boroxine K2[B3O3F4OH] (HB), an inorganic derivative of cyclic anhydride of boronic acid, is patented as a boron-containing compound with potential for the treatment of both benign and malignant skin changes. HB has effectively inhibited the growth of several carcinoma cell lines. Because of the growing interest in autophagy induction as a therapeutic approach in bladder carcinoma (BC), we aimed to assess the effects of HB on metabolic phenotype and autophagy levels in 5637 human bladder carcinoma cells (BC). Cytotoxicity was evaluated using the alamar blue assay, and the degree of autophagy was determined microscopically. Mitochondrial respiration and glycolysis were measured simultaneously. The relative expression of autophagy-related genes BECN1, P62, BCL-2, and DRAM1 was determined by real-time PCR. HB affected cell growth, while starvation significantly increased the level of autophagy in the positive control compared to the basal level of autophagy in the untreated negative control. In HB-treated cultures, the degree of autophagy was higher compared to the basal level, and metabolic phenotypes were altered; both glycolysis and oxidative phosphorylation (OXPHOS) were decreased by HB at 0.2 and 0.4 mg/mL. Gene expression was deregulated towards autophagy induction and expansion. In conclusion, HB disrupted the bioenergetic metabolism and reduced the intracellular survival potential of BC cells. Further molecular studies are needed to confirm these findings and investigate their applicative potential.


Subject(s)
Autophagy , Urinary Bladder Neoplasms , Humans , Autophagy/drug effects , Cell Line, Tumor , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/genetics , Cell Proliferation/drug effects , Glycolysis/drug effects , Phenotype , Oxidative Phosphorylation/drug effects , Cell Survival/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Halogenation
19.
Cell Commun Signal ; 22(1): 336, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898530

ABSTRACT

Excessive scar formation such as hypertrophic scars and keloids, resulting from trauma or surgical procedures, present a widespread concern for causing disfigurement, discomfort, and functional limitations. Macrophages play pivotal roles in maintaining tissue homeostasis, orchestrating tissue development, repair, and immune responses, and its transition of function and phenotype plays a critical role in regulating the balance between inflammation and tissue regeneration, which is central to cutaneous scar formation. Recent evidence suggests the involvement of Sonic Hedgehog (SHH) in the induction of anti-inflammatory M2-like macrophage phenotypes within tumor microenvironments. In our study, we observed increased SHH expression in human hypertrophic scars, prompting an investigation into its influence on macrophage polarization, efferocytosis, and cutaneous scar formation. Our findings reveal that SHH can enhance oxidative phosphorylation (OXPHOS) in macrophages, augment macrophage efferocytosis, and promote M2 polarization, finally contributing to the progression of cutaneous scar formation. Notably, targeting SHH signaling with vismodegib exhibited promising potential in mitigating scar formation by reversing the effects of enhanced OXPHOS and M2 polarization in macrophages. In conclusion, this study underscores the critical roles of macrophage metabolism, particularly OXPHOS, efferocytosis and SHH signaling in cutaneous scar formation. Understanding these mechanisms provides new avenues for potential interventions and scar prevention strategies.


Subject(s)
Hedgehog Proteins , Macrophages , Oxidative Phosphorylation , Phagocytosis , Hedgehog Proteins/metabolism , Macrophages/metabolism , Macrophages/drug effects , Humans , Oxidative Phosphorylation/drug effects , Animals , Phagocytosis/drug effects , Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/pathology , Mice , Signal Transduction/drug effects , Cicatrix/pathology , Cicatrix/metabolism , Mice, Inbred C57BL , Anilides/pharmacology , Pyridines/pharmacology , Efferocytosis
20.
J Transl Med ; 22(1): 535, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38840216

ABSTRACT

BACKGROUND: Inflammation and endothelial barrier dysfunction are the major pathophysiological changes in acute respiratory distress syndrome (ARDS). Sphingosine-1-phosphate receptor 3 (S1PR3), a G protein-coupled receptor, has been found to mediate inflammation and endothelial cell (EC) integrity. However, the function of S1PR3 in ARDS has not been fully elucidated. METHODS: We used a murine lipopolysaccharide (LPS)-induced ARDS model and an LPS- stimulated ECs model to investigate the role of S1PR3 in anti-inflammatory effects and endothelial barrier protection during ARDS. RESULTS: We found that S1PR3 expression was increased in the lung tissues of mice with LPS-induced ARDS. TY-52156, a selective S1PR3 inhibitor, effectively attenuated LPS-induced inflammation by suppressing the expression of proinflammatory cytokines and restored the endothelial barrier by repairing adherens junctions and reducing vascular leakage. S1PR3 inhibition was achieved by an adeno-associated virus in vivo and a small interfering RNA in vitro. Both the in vivo and in vitro studies demonstrated that pharmacological or genetic inhibition of S1PR3 protected against ARDS by inhibiting the NF-κB pathway and improving mitochondrial oxidative phosphorylation. CONCLUSIONS: S1PR3 inhibition protects against LPS-induced ARDS via suppression of pulmonary inflammation and promotion of the endothelial barrier by inhibiting NF-κB and improving mitochondrial oxidative phosphorylation, indicating that S1PR3 is a potential therapeutic target for ARDS.


Subject(s)
Lipopolysaccharides , Mice, Inbred C57BL , Mitochondria , NF-kappa B , Oxidative Phosphorylation , Respiratory Distress Syndrome , Sphingosine-1-Phosphate Receptors , Animals , Humans , Male , Mice , Cytokines/metabolism , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Inflammation/pathology , Lung/pathology , Lung/drug effects , Lung/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , NF-kappa B/metabolism , Oxidative Phosphorylation/drug effects , Protective Agents/pharmacology , Receptors, Lysosphingolipid/metabolism , Receptors, Lysosphingolipid/antagonists & inhibitors , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL